Ursodeoxycholate (BioDeep_00000000066)

 

Secondary id: BioDeep_00000229700

human metabolite Endogenous blood metabolite Bile acids PANOMIX LipidSearch


代谢物信息卡片


(4R)-4-[(1S,2S,5R,7S,9S,10R,11S,14R,15R)-5,9-dihydroxy-2,15-dimethyltetracyclo[8.7.0.0^{2,7}.0^{11,15}]heptadecan-14-yl]pentanoic acid

化学式: C24H40O4 (392.292644)
中文名称: 熊去氧胆酸, 熊脱氧胆酸
谱图信息: 最多检出来源 Homo sapiens(bile_acids) 0.09%

Reviewed

Last reviewed on 2024-09-13.

Cite this Page

Ursodeoxycholate. BioDeep Database v3. PANOMIX ltd, a top metabolomics service provider from China. https://query.biodeep.cn/s/ursodeoxycholate (retrieved 2024-09-17) (BioDeep RN: BioDeep_00000000066). Licensed under the Attribution-Noncommercial 4.0 International License (CC BY-NC 4.0).

分子结构信息

SMILES: C[C@H](CCC(=O)O)[C@H]1CC[C@H]2[C@H]3[C@H](CC[C@]12C)[C@@]1(C)CC[C@H](C[C@H]1C[C@@H]3O)O
InChI: InChI=1S/C24H40O4/c1-14(4-7-21(27)28)17-5-6-18-22-19(9-11-24(17,18)3)23(2)10-8-16(25)12-15(23)13-20(22)26/h14-20,22,25-26H,4-13H2,1-3H3,(H,27,28)/t14-,15?,16+,17-,18+,19+,20+,22+,23+,24-/m1/s1

描述信息

Ursodeoxycholic acid is a bile acid found in the bile of bears (Ursidae) as a conjugate with taurine. Used therapeutically, it prevents the synthesis and absorption of cholesterol and can lead to the dissolution of gallstones. It has a role as a human metabolite and a mouse metabolite. It is a bile acid, a dihydroxy-5beta-cholanic acid and a C24-steroid. It is a conjugate acid of an ursodeoxycholate.
Ursodeoxycholic acid is an epimer of [chenodeoxycholic acid]. It is a mammalian bile acid found first in the bear and is apparently either a precursor or a product of chenodeoxycholate. Its administration changes the composition of bile and may dissolve gallstones. It is used as a cholagogue and choleretic.
Ursodiol is a Bile Acid.
Ursodeoxycholic acid or ursodiol is a naturally occurring bile acid that is used dissolve cholesterol gall stones and to treat cholestatic forms of liver diseases including primary biliary cirrhosis. Ursodiol has been linked to rare instances of transient and mild serum aminotransferase elevations during therapy and to rare instances of jaundice and worsening of liver disease in patients with preexisting cirrhosis.
Ursodeoxycholic acid is a natural product found in Myocastor coypus with data available.
Ursodiol is a synthetically-derived form of ursodiol, a bile acid produced by the liver and secreted and stored in the gallbladder. Also produced by the Chinese black bear liver, ursodiol has been used in the treatment of liver disease for centuries. This agent dissolves or prevents cholesterol gallstones by blocking hepatic cholesterol production and decreasing bile cholesterol. Ursodiol also reduces the absorption of cholesterol from the intestinal tract.
An epimer of chenodeoxycholic acid. It is a mammalian bile acid found first in the bear and is apparently either a precursor or a product of chenodeoxycholate. Its administration changes the composition of bile and may dissolve gallstones. It is used as a cholagogue and choleretic.
See also: Dimethicone; pancrelipase; ursodiol (component of).
Ursodeoxycholic acid, also known as ursodeoxycholate or acid deoxyursocholic, belongs to the class of organic compounds known as dihydroxy bile acids, alcohols and derivatives. Dihydroxy bile acids, alcohols and derivatives are compounds containing or derived from a bile acid or alcohol, and which bears exactly two carboxylic acid groups. Ursodeoxycholic acid is a very hydrophobic molecule, practically insoluble in water, and relatively neutral.
An epimer of chenodeoxycholic acid. It is a mammalian bile acid found first in the bear and is apparently either a precursor or a product of chenodeoxycholate. Its administration changes the composition of bile and may dissolve gallstones. It is used as a cholagogue and choleretic. [HMDB]

Ursodeoxycholic acid. CAS Common Chemistry. CAS, a division of the American Chemical Society, n.d. https://commonchemistry.cas.org/detail?cas_rn=128-13-2 (retrieved 2024-07-02) (CAS RN: 128-13-2). Licensed under the Attribution-Noncommercial 4.0 International License (CC BY-NC 4.0).

同义名列表

181 个代谢物同义名

(4R)-4-[(3R,5S,7S,8R,9S,10S,13R,14S,17R)-10,13-dimethyl-3,7-bis(oxidanyl)-2,3,4,5,6,7,8,9,11,12,14,15,16,17-tetradecahydro-1H-cyclopenta[a]phenanthren-17-yl]pentanoic acid; (4R)-4-[(3R,5S,7S,8R,9S,10S,13R,14S,17R)-3,7-dihydroxy-10,13-dimethyl-2,3,4,5,6,7,8,9,11,12,14,15,16,17-tetradecahydro-1H-cyclopenta[a]phenanthren-17-yl]pentanoic acid; (4R)-4-[(3R,5S,7S,8R,9S,10S,13R,14S,17R)-3,7-dihydroxy-10,13-dimethyl-2,3,4,5,6,7,8,9,11,12,14,15,16,17-tetradecahydro-1H-cyclopenta[a]phenanthren-17-yl]pentanoicacid; (4R)-4-[(3R,5S,7S,8R,9S,10S,13R,14S,17R)-3,7-dihydroxy-10,13-dimethyl-2,3,4,5,6,7,8,9,11,12,14,15,16,17-tetradecahydro-1H-cyclopenta[a]phenanthren-17-yl]valeric acid; (4R)-4-[(1R,3aS,3bR,4S,5aS,7R,9aS,9bS,11aR)-4,7-dihydroxy-9a,11a-dimethyl-hexadecahydro-1H-cyclopenta[a]phenanthren-1-yl]pentanoic acid; (R)-4-((3R,5S,7S,8R,9S,10S,13R,14S,17R)-3,7-dihydroxy-10,13-dimethylhexadecahydro-1H-cyclopenta[a]phenanthren-17-yl)pentanoic acid; (4R)-4-[(1S,2S,5R,7S,9S,10R,11S,14R,15R)-5,9-Dihydroxy-2,15-dimethyltetracyclo[8.7.0.02,1.011,1]heptadecan-14-yl]pentanoic acid; Ursodeoxycholic acid for system suitability, European Pharmacopoeia (EP) Reference Standard; Ursodiol, Pharmaceutical Secondary Standard; Certified Reference Material; 17.beta.-(1-Methyl-3-carboxypropyl)etiocholane-3.alpha.,7.beta.-diol; Ursodeoxycholic acid, European Pharmacopoeia (EP) Reference Standard; Ursodeoxycholic acid, British Pharmacopoeia (BP) Reference Standard; 17-beta-(1-Methyl-3-carboxypropyl)etiocholane-3-alpha,7-beta-diol; 3,7-Dihydroxycholan-24-oic acid-, (3.alpha.,5.beta.,7.beta.)- #; Cholan-24-oic acid, 3,7-dihydroxy-, (3.alpha.,5.beta.,7.beta.)-; Ursodiol, United States Pharmacopeia (USP) Reference Standard; Cholan-24-oic acid, 3,7-dihydroxy-, (3-alpha,5-beta,7-beta)-; Cholan-24-oic acid, 3,7-dihydroxy-, (3alpha,5beta,7beta)-; (3alpha,5beta,7beta,8xi)-3,7-dihydroxycholan-24-oic acid; 5.beta.-Cholan-24-oic acid, 3.alpha.,7.beta.-dihydroxy-; 3.alpha.,7.beta.-Dihydroxy-5.beta.-cholan-24-oic acid; (3alpha,5beta,7beta)-3,7-dihydroxycholan-24-oic acid; 5-beta-Cholan-24-oic acid, 3-alpha,7-beta-dihydroxy-; 3 alpha,7 beta-Dihydroxy-5 beta-cholan-24-oic Acid; 3 alpha,7 beta Dihydroxy 5 beta cholan 24 oic Acid; (3alpha,5beta,7beta)-3,7-Dihydroxycholan-24-Oate; 3.alpha.,7.beta.-Dihydroxy-5.beta.-cholanic acid; 3.ALPHA.,7.BETA.-DIHYDROXY-5B-CHOLAN-24-OIC ACID; Sanofi synthelabo brand OF ursodeoxycholic acid; 3alpha,7beta-Dihydroxy-5beta-cholan-24-oic acid; 3-alpha,7-beta-Dihydroxy-5-beta-cholanoic acid; 4-10-00-01604 (Beilstein Handbook Reference); 3alpha,7beta-DIHYDROXY-5B-CHOLAN-24-OIC ACID; 3alpha, 7beta-dihydroxy-5beta-cholanoic acid; 3alpha,7beta-Dihydroxy-5beta-cholan-24-Oate; ((3a,5b,7b)-3,7-Dihydroxycholan-24-oic acid; (3a,5b,7b)-3,7-dihydroxy-cholan-24-oic acid; (3a,5b,7b)-3,7-dihydroxycholan-24-oic acid; (3Α,5β,7β)-3,7-dihydroxycholan-24-Oic acid; 3alpha,7beta-Dihydroxy-5beta-cholanic acid; Niddapharm brand OF ursodeoxycholic acid; (3a,5b,7b)-3,7-dihydroxy-cholan-24-oate; 3.alpha.,7.beta.-Dihydroxycholanic acid; Tramedico brand OF ursodeoxycholic acid; Provalis brand OF ursodeoxycholic acid; (3a,5b,7b)-3,7-Dihydroxycholan-24-Oate; (3Α,5β,7β)-3,7-dihydroxycholan-24-Oate; Farmasa brand OF ursodeoxycholic acid; 3Α,7β-dihydroxy-5β-cholan-24-Oic acid; 5beta-Cholanic Acid-3alpha,7beta-diol; Norgine brand OF ursodeoxycholic acid; Acide ursodesoxycholique (INN-French); 3a,7b-dihydroxy-5b-cholan-24-oic acid; Heumann brand OF ursodeoxycholic acid; Aventis brand OF ursodeoxycholic acid; Acide ursodesoxycholique [INN-French]; Antigen brand OF ursodeoxycholic acid; 3-alpha,7-beta-Dihydroxycholanic acid; Zambon brand OF ursodeoxycholic acid; Acidum ursodeoxycholicum (INN-Latin); Estedi brand OF ursodeoxycholic acid; Acidum ursodeoxycholicum [INN-Latin]; 3A,7A-Dihydroxy-5A-holan-24-oic acid; Orphan brand OF ursodeoxycholic acid; Acido ursodeoxicolico (INN-Spanish); URSODEOXYCHOLIC ACID [EP MONOGRAPH]; URSODEOXYCHOLIC ACID (EP MONOGRAPH); Galen brand OF ursodeoxycholic acid; Acido ursodeoxicolico [INN-Spanish]; Axcan brand OF ursodeoxycholic acid; 3alpha,7beta-dihydroxycholanic acid; Falk brand OF ursodeoxycholic acid; Vita brand OF ursodeoxycholic acid; URSODEOXYCHOLIC ACID [EP IMPURITY]; URSODEOXYCHOLIC ACID (EP IMPURITY); 3a,7b-dihydroxy-5b-cholan-24-oate; 3Α,7β-dihydroxy-5β-cholan-24-Oate; 3-alpha,7-beta-Dioxycholanic acid; CP Brand OF ursodeoxycholic acid; Acido ursodeossicolico [Italian]; 5A-Cholan-24-oic acid-3A,7A-diol; Ursodeoxycholic acid (JP17/INN); 3,7-Dihydroxycholan-24-oic acid; 7.beta.-Hydroxylithocholic acid; Ursodeoxycholic Acid (Ursodiol); 7-beta-Hydroxylithocholic acid; URSODEOXYCHOLIC ACID [WHO-DD]; URSODEOXYCHOLIC ACID [MART.]; URSODEOXYCHOLIC ACID (MART.); Ursodeoxycholic acid, >=99\\%; 7A-Hydroxylithocholic acid; URSODEOXYCHOLIC ACID [JAN]; Ursodeoxycholic acid [INN]; acidum ursodesoxycholicum; Acide ursodesoxycholique; Ursodeoxycholate, Sodium; URSODIOL (USP MONOGRAPH); URSODIOL [USP MONOGRAPH]; Acidum ursodeoxycholicum; Ursocholic acid, deoxy-; Ursodiol (Actigal Urso); acide ursodeoxycholique; acido ursodesossicolico; Sodium Ursodeoxycholate; URSODIOL [ORANGE BOOK]; Acido ursodeossicolico; Acid, Ursodeoxycholic; Ursodesoxycholic acid; Acido ursodeoxicolico; Acid, Deoxyursocholic; Urosdesoxycholic acid; Ursodeoxycholic Acid; Ursodeoxycholoc acid; Deoxyursocholic Acid; Ursodexycholic Acid; Ursodeoxycholicacid; Ursodiol [USAN:USP]; Ursodiol (USAN:USP); Prestwick0_000958; URSODIOL [USP-RS]; URSODIOL (USP-RS); Prestwick3_000958; Urosdesoxycholate; Prestwick1_000958; Prestwick2_000958; ursodeoxycholate; Acid, Ursacholic; URSODIOL [VANDF]; Ursodeoxycholic; Ursacholic Acid; URSODIOL [INCI]; Ursodexycholate; Ursodiol [USAN]; Ursodiol (USP); Actigall (TN); BPBio1_001052; URSODIOL [MI]; Urso Heumann; Cholit-ursan; NCI60_028904; SMP2_000012; Ursobilane; ST 24:1;O4; Urso Forte; Urso (TN); Ursobilin; Ursodamor; Cholofalk; Ursolite; Actigall; Litursol; Peptarom; Ursodiol; Ursolvan; Solutrat; Destolit; Ursofalk; Antigall; Delursan; Ursonorm; Paptarom; Ursochol; Ursogal; Deursil; Usodiol; Arsacol; A05AA02; Ursacol; Urosiol; Ursosan; Urso DS; Lyeton; Udiliv; URUSA; Urdes; Desol; Urdox; Urso; UDCS; UDCA; Ursodeoxycholic acid (UDCA)



数据库引用编号

26 个数据库交叉引用编号

分类词条

相关代谢途径

Reactome(0)

BioCyc(0)

PlantCyc(0)

代谢反应

0 个相关的代谢反应过程信息。

Reactome(0)

BioCyc(0)

WikiPathways(0)

Plant Reactome(0)

INOH(0)

PlantCyc(0)

COVID-19 Disease Map(0)

PathBank(0)

PharmGKB(0)

3 个相关的物种来源信息

在这里通过桑基图来展示出与当前的这个代谢物在我们的BioDeep知识库中具有相关联信息的其他代谢物。在这里进行关联的信息来源主要有:

  • PubMed: 来源于PubMed文献库中的文献信息,我们通过自然语言数据挖掘得到的在同一篇文献中被同时提及的相关代谢物列表,这个列表按照代谢物同时出现的文献数量降序排序,取前10个代谢物作为相关研究中关联性很高的代谢物集合展示在桑基图中。
  • NCBI Taxonomy: 通过文献数据挖掘,得到的代谢物物种来源信息关联。这个关联信息同样按照出现的次数降序排序,取前10个代谢物作为高关联度的代谢物集合展示在桑吉图上。
  • Chemical Taxonomy: 在物质分类上处于同一个分类集合中的其他代谢物
  • Chemical Reaction: 在化学反应过程中,存在为当前代谢物相关联的生化反应过程中的反应底物或者反应产物的关联代谢物信息。

点击图上的相关代谢物的名称,可以跳转到相关代谢物的信息页面。



文献列表

  • Rulaiha Taylor, Zhenning Yang, Zakiyah Henry, Gina Capece, Vik Meadows, Katherine Otersen, Veronia Basaly, Anisha Bhattacharya, Stephanie Mera, Peihong Zhou, Laurie Joseph, Ill Yang, Anita Brinker, Brian Buckley, Bo Kong, Grace L Guo. Characterization of individual bile acids in vivo utilizing a novel low bile acid mouse model. Toxicological sciences : an official journal of the Society of Toxicology. 2024 May; 199(2):316-331. doi: 10.1093/toxsci/kfae029. [PMID: 38526215]
  • Shu Dai, Rui Wu, Ke Fu, Yanzhi Li, Chenghao Yao, Yanfang Liu, Fang Zhang, Shenglin Zhang, Yiling Guo, Yuxin Yao, Yunxia Li. Exploring the effect and mechanism of cucurbitacin B on cholestatic liver injury based on network pharmacology and experimental verification. Journal of ethnopharmacology. 2024 Mar; 322(?):117584. doi: 10.1016/j.jep.2023.117584. [PMID: 38104874]
  • Yanbo Chang, Xuejing Li, Jinping Jiang, Lanlan Gui, Linfei Wan, Xiangxiang Zhou, Linchuan Liao, Kexin Li, Ke Lan. Separation of bile acid isomer plays a pivotal role in bioequivalence evaluation of ursodeoxycholic acid. Journal of pharmaceutical and biomedical analysis. 2024 Feb; 239(?):115882. doi: 10.1016/j.jpba.2023.115882. [PMID: 38071766]
  • Jianqing She, Gulinigaer Tuerhongjiang, Manyun Guo, Junhui Liu, Xiang Hao, Liangan Guo, Nairong Liu, Wen Xi, Tao Zheng, Bin Du, Bowen Lou, Xiyu Gao, Xiao Yuan, Yue Yu, Yi Zhang, Fan Gao, Xiaozhen Zhuo, Ying Xiong, Xiang Zhang, Jun Yu, Zuyi Yuan, Yue Wu. Statins aggravate insulin resistance through reduced blood glucagon-like peptide-1 levels in a microbiota-dependent manner. Cell metabolism. 2024 02; 36(2):408-421.e5. doi: 10.1016/j.cmet.2023.12.027. [PMID: 38325336]
  • Biljana Lakić, Ranko Škrbić, Snežana Uletilović, Nebojša Mandić-Kovačević, Milkica Grabež, Mirna Popović Šarić, Miloš P Stojiljković, Ivan Soldatović, Zorica Janjetović, Anastasija Stokanović, Nataša Stojaković, Momir Mikov. Beneficial Effects of Ursodeoxycholic Acid on Metabolic Parameters and Oxidative Stress in Patients with Type 2 Diabetes Mellitus: A Randomized Double-Blind, Placebo-Controlled Clinical Study. Journal of diabetes research. 2024; 2024(?):4187796. doi: 10.1155/2024/4187796. [PMID: 38455850]
  • Yu-Qiong He, Jiu-Ling Deng, Can-Can Zhou, Sheng-Gui Jiang, Feng Zhang, Xia Tao, Wan-Sheng Chen. Ursodeoxycholic acid alleviates sepsis-induced lung injury by blocking PANoptosis via STING pathway. International immunopharmacology. 2023 Dec; 125(Pt B):111161. doi: 10.1016/j.intimp.2023.111161. [PMID: 37948864]
  • Xiaoxue Li, Yue Hu, Bingxin He, Lingyu Li, Yu Tian, Yingjie Xiao, Hai Shang, Zhongmei Zou. Design, synthesis and evaluation of ursodeoxycholic acid-cinnamic acid hybrids as potential anti-inflammatory agents by inhibiting Akt/NF-κB and MAPK signaling pathways. European journal of medicinal chemistry. 2023 Nov; 260(?):115785. doi: 10.1016/j.ejmech.2023.115785. [PMID: 37678142]
  • Fei Qiao, Feng Ren, Weiting Lu, Haoran Yang, Guiling Mo, Shuangshuang Wang, Lina Liu, Xiangtao Xu. A female of progressive familial intrahepatic cholestasis type 3 caused by heterozygous mutations of ABCB4 gene and her cirrhosis improved after treatment of ursodeoxycholic acid: a case report. BMC medical genomics. 2023 07; 16(1):171. doi: 10.1186/s12920-023-01602-y. [PMID: 37488596]
  • Wei Li, Wei Chen, Xiaoya Niu, Chen Zhao, Pengfei Tu, Jun Li, Wenjing Liu, Yuelin Song. Characterization of Metabolic Correlations of Ursodeoxycholic Acid with Other Bile Acid Species through In Vitro Sequential Metabolism and Isomer-Focused Identification. Molecules (Basel, Switzerland). 2023 Jun; 28(12):. doi: 10.3390/molecules28124801. [PMID: 37375356]
  • M A Rasskazova, S V Vorobyev, H N Butova. [Possibilities for the use of ursodeoxycholic acid in the treatment of patients with type 2 diabetes mellitus and non-alcoholic fatty liver disease]. Terapevticheskii arkhiv. 2023 May; 95(4):316-321. doi: 10.26442/00403660.2023.04.202125. [PMID: 38158979]
  • Shan Gao, Feng Gao, Jing-Wei Kong, Zhi-Jia Wang, Hao-Cheng Zheng, Xin-Qi Jiang, Shu-Jing Xu, Shan-Lan Li, Ming-Jun Lu, Zi-Qi Dai, Fu-Hao Chu, Bing Xu, Hai-Min Lei. [Therapeutic effect of ursodeoxycholic acid-berberine supramolecular nanoparticles on ulcerative colitis based on supramolecular system induced by weak bond]. Zhongguo Zhong yao za zhi = Zhongguo zhongyao zazhi = China journal of Chinese materia medica. 2023 May; 48(10):2739-2748. doi: 10.19540/j.cnki.cjcmm.20230223.301. [PMID: 37282934]
  • Jingfeng Yu, Sihan Sheng, Xiaosu Zou, Zhengwu Shen. Dihydroartemisinin-ursodeoxycholic acid conjugate is a potential treatment agent for inflammatory bowel disease. International immunopharmacology. 2023 Feb; 117(?):109918. doi: 10.1016/j.intimp.2023.109918. [PMID: 36842236]
  • Silvia Marchianò, Michele Biagioli, Elva Morretta, Cristina Di Giorgio, Rosalinda Roselli, Martina Bordoni, Rachele Bellini, Ginevra Urbani, Carmen Massa, Maria Chiara Monti, Angela Zampella, Eleonora Distrutti, Stefano Fiorucci. Combinatorial therapy with BAR502 and UDCA resets FXR and GPBAR1 signaling and reverses liver histopathology in a model of NASH. Scientific reports. 2023 Jan; 13(1):1602. doi: 10.1038/s41598-023-28647-4. [PMID: 36709356]
  • Haolong Li, Haoting Zhan, Linlin Cheng, Yuan Huang, Xiaomeng Li, Songxin Yan, Yongmei Liu, Li Wang, Yongzhe Li. Plasma lipidomics of primary biliary cholangitis and its comparison with Sjögren's syndrome. Frontiers in immunology. 2023; 14(?):1124443. doi: 10.3389/fimmu.2023.1124443. [PMID: 37215104]
  • Shiori Ishizawa, Akinori Nishi, Noriko Kaifuchi, Chika Shimobori, Miwa Nahata, Chihiro Yamada, Seiichi Iizuka, Katsuya Ohbuchi, Mitsue Nishiyama, Naoki Fujitsuka, Toru Kono, Masahiro Yamamoto. Integrated analysis of effect of daisaikoto, a traditional Japanese medicine, on the metabolome and gut microbiome in a mouse model of nonalcoholic fatty liver disease. Gene. 2022 Dec; 846(?):146856. doi: 10.1016/j.gene.2022.146856. [PMID: 36067864]
  • Silvia Marchianò, Michele Biagioli, Rosalinda Roselli, Angela Zampella, Cristina Di Giorgio, Martina Bordoni, Rachele Bellini, Ginevra Urbani, Elva Morretta, Maria Chiara Monti, Eleonora Distrutti, Stefano Fiorucci. Beneficial effects of UDCA and norUDCA in a rodent model of steatosis are linked to modulation of GPBAR1/FXR signaling. Biochimica et biophysica acta. Molecular and cell biology of lipids. 2022 11; 1867(11):159218. doi: 10.1016/j.bbalip.2022.159218. [PMID: 35985473]
  • Ting Gong, Chuangen Li, Shiqiang Li, Xiaojuan Zhang, Zhongming He, Xianhong Jiang, Qiuyue He, Rongjuan Huang, Yong Wang, Xiong Liu. Capsaicin regulates dyslipidemia by altering the composition of bile acids in germ-free mice. Food & function. 2022 Oct; 13(20):10665-10679. doi: 10.1039/d2fo02209e. [PMID: 36172720]
  • Da-Xin Chen, Jian-Feng Chu, Shan Lin, Ling Zhang, Hong-Wei Chen, Zhi-Wei Sun, Jian-Feng Xu, Qiao-Yan Cai, Li-Li Wang, Jun Peng. Therapeutic Effects of Different Animal Bile Powders on Lipid Metabolism Disorders and Their Composition Analysis. Chinese journal of integrative medicine. 2022 Oct; 28(10):918-923. doi: 10.1007/s11655-021-3441-3. [PMID: 33609233]
  • Haining Peng, Yi Zhang, Zhongkai Ren, Ziran Wei, Renjie Chen, Yingze Zhang, Xiaohong Huang, Tengbo Yu. Cartilaginous Metabolomics Reveals the Biochemical-Niche Fate Control of Bone Marrow-Derived Stem Cells. Cells. 2022 09; 11(19):. doi: 10.3390/cells11192951. [PMID: 36230915]
  • Rumei Li, Milaine V Hovingh, Martijn Koehorst, Pim de Blaauw, Henkjan J Verkade, Jan Freark de Boer, Folkert Kuipers. Short-term obeticholic acid treatment does not impact cholangiopathy in Cyp2c70-deficient mice with a human-like bile acid composition. Biochimica et biophysica acta. Molecular and cell biology of lipids. 2022 08; 1867(8):159163. doi: 10.1016/j.bbalip.2022.159163. [PMID: 35470044]
  • Surain B Roberts, Gideon M Hirschfield, Lawrence J Worobetz, Catherine Vincent, Jennifer A Flemming, Angela Cheung, Karim Qumosani, Mark Swain, Dusanka Grbic, Hin Hin Ko, Kevork Peltekian, Nazia Selzner, Lusine Abrahamyan, Bishoi Aziz, Ellina Lytvyak, Kattleya Tirona, Aliya F Gulamhusein, Harry L A Janssen, Aldo J Montano-Loza, Andrew L Mason, Bettina E Hansen. Ethnicity, disease severity, and survival in Canadian patients with primary biliary cholangitis. Hepatology (Baltimore, Md.). 2022 Aug; 76(2):303-316. doi: 10.1002/hep.32426. [PMID: 35220609]
  • Holly Lovell, Alice Mitchell, Caroline Ovadia, Noelia Pitrelli, Annette Briley, Claire Singh, Hanns-Ulrich Marschall, Kennedy Cruickshank, Helen Murphy, Paul Seed, Catherine Williamson. A multi-centered trial investigating gestational treatment with ursodeoxycholic acid compared to metformin to reduce effects of diabetes mellitus (GUARD): a randomized controlled trial protocol. Trials. 2022 Jul; 23(1):571. doi: 10.1186/s13063-022-06462-y. [PMID: 35854327]
  • Pengchao Xie, Yan Peng, Liyan Qiu. Responsive oligochitosan nano-vesicles with ursodeoxycholic acid and exenatide for NAFLD synergistic therapy via SIRT1. Carbohydrate polymers. 2022 Jul; 288(?):119388. doi: 10.1016/j.carbpol.2022.119388. [PMID: 35450649]
  • Yaping Guan, Fei Xu, Xiaodong Zhang, Xiao Fu, Jing Wang, Sentao Song, Yan Sun, Qiongying Yuan, Feng Zhu. Roles of ursodeoxycholic acid in the bile biochemistry and metabolomics in patients with choledocholithiasis: a prospective study. Metabolomics : Official journal of the Metabolomic Society. 2022 07; 18(7):46. doi: 10.1007/s11306-022-01906-7. [PMID: 35778620]
  • Pham Xuan Thuy, Tran Duc Duy Bao, Eun-Yi Moon. Ursodeoxycholic acid ameliorates cell migration retarded by the SARS-CoV-2 spike protein in BEAS-2B human bronchial epithelial cells. Biomedicine & pharmacotherapy = Biomedecine & pharmacotherapie. 2022 Jun; 150(?):113021. doi: 10.1016/j.biopha.2022.113021. [PMID: 35658221]
  • Dawei Ding, Guoyun Xuan, Yinan Hu, Jiahao Yu, Yansheng Liu, Guanya Guo, Shuoyi Ma, Fangfang Yang, Siyuan Tian, Gang Ma, Ling Chen, Xinmin Zhou, Yulong Shang, Ying Han. Immunoglobulin M: A Neglected Serum Biomarker in Treatment-Naive Primary Biliary Cholangitis With Normal Alkaline Phosphatase. Hepatology communications. 2022 Jun; 6(6):1403-1412. doi: 10.1002/hep4.1907. [PMID: 35182047]
  • Tamás Deli, Judit Tóth, Lea Csépes-Ruzicska, Olga Török, Zoárd Tibor Krasznai, Ágnes Mosolygó-Lukács, János Kappelmayer, Rudolf Lampé. Az epesavmérés szerepe a súlyos terhességi cholestasis szülészeti ellátásában. Orvosi hetilap. 2022 May; 163(20):797-805. doi: 10.1556/650.2022.32474. [PMID: 35569059]
  • M V Maevskaya, Y V Kotovskaya, V T Ivashkin, O N Tkacheva, E A Troshina, M V Shestakova, V V Breder, N I Geyvandova, V L Doshchitsin, E N Dudinskaya, E V Ershova, K B Kodzoeva, K A Komshilova, N V Korochanskaya, A Y Mayorov, E E Mishina, M Y Nadinskaya, I G Nikitin, N V Pogosova, A I Tarzimanova, M S Shamkhalova. [The National Consensus statement on the management of adult patients with non-alcoholic fatty liver disease and main comorbidities]. Terapevticheskii arkhiv. 2022 Feb; 94(2):216-253. doi: 10.26442/00403660.2022.02.201363. [PMID: 36286746]
  • Weijia Han, Chunyang Huang, Qi Zhang, Shuhui Tao, Xiaomin Hu, Jianguo Xu, Ronglong Jiang, Bin Xu, Yanmin Liu, Jinlin Hou. Alterations in gut microbiota and elevated serum bilirubin in primary biliary cholangitis patients treated with ursodeoxycholic acid. European journal of clinical investigation. 2022 Feb; 52(2):e13714. doi: 10.1111/eci.13714. [PMID: 34800290]
  • Si-Ying Liu, Li-Wen Chang, Jing Wang, Min Xie, Lei-Lei Chen, Wei Liu. Ursodeoxycholic acid prevention on cholestasis associated with total parenteral nutrition in preterm infants: a randomized trial. World journal of pediatrics : WJP. 2022 02; 18(2):100-108. doi: 10.1007/s12519-021-00487-0. [PMID: 34988851]
  • Paul J McMurdie, Magdalena K Stoeva, Nicholas Justice, Madeleine Nemchek, Christian M K Sieber, Surabhi Tyagi, Jessica Gines, Connor T Skennerton, Michael Souza, Orville Kolterman, John Eid. Increased circulating butyrate and ursodeoxycholate during probiotic intervention in humans with type 2 diabetes. BMC microbiology. 2022 01; 22(1):19. doi: 10.1186/s12866-021-02415-8. [PMID: 34996347]
  • Lin Li, Yi Song, Xinyu Chen, Yi Huang, Huabao Liu. [Bioinformatic analysis of differentially expressed genes and immune cell infiltration in primary biliary cholangitis patients with poor response to ursodeoxycholic acid therapy]. Xi bao yu fen zi mian yi xue za zhi = Chinese journal of cellular and molecular immunology. 2022 Jan; 38(1):16-23. doi: . [PMID: 35078571]
  • Armin Mooranian, Nassim Zamani, Bozica Kovacevic, Corina Mihaela Ionescu, Giuseppe Luna, Momir Mikov, Svetlana Goločorbin-Kon, Goran Stojanovic, Sanja Kojic, Hani Al-Salami. Pharmacological Effects of Secondary Bile Acid Microparticles in Diabetic Murine Model. Current diabetes reviews. 2022; 18(1):e062620183199. doi: 10.2174/1573399816666200626213735. [PMID: 32589561]
  • Hajime Ueda, Akira Honda, Teruo Miyazaki, Yukio Morishita, Takeshi Hirayama, Junichi Iwamoto, Nobuhiro Nakamoto, Tadashi Ikegami. Sex-, age-, and organ-dependent improvement of bile acid hydrophobicity by ursodeoxycholic acid treatment: A study using a mouse model with human-like bile acid composition. PloS one. 2022; 17(7):e0271308. doi: 10.1371/journal.pone.0271308. [PMID: 35819971]
  • Lingmei Zeng, Mao Li, Yuting Cen, Zuanyu Wang. Effects of S-adenosyl-L-Methionine Combined with Ursodesoxycholic Acid on Serum Endotoxin, MMP-9 and IL-18 in Neonates with Cholestasis. Journal of the College of Physicians and Surgeons--Pakistan : JCPSP. 2021 Dec; 31(12):1445-1448. doi: 10.29271/jcpsp.2021.12.1445. [PMID: 34794285]
  • A Fouda, A E Abdelaziz, M Hussien, A A Ali, K S Abdelkawy, F Elbarbry. A randomized controlled trial comparing the effects of Vitamin E, Ursodeoxycholic acid and Pentoxifylline on Egyptian non-alcoholic steatohepatitis patients. European review for medical and pharmacological sciences. 2021 Dec; 25(23):7449-7459. doi: 10.26355/eurrev_202112_27442. [PMID: 34919247]
  • Ben Barron-Millar, Laura Ogle, George Mells, Steven Flack, Jonathan Badrock, Richard Sandford, John Kirby, Jeremy Palmer, Laura Jopson, John Brain, Graham R Smith, Steve Rushton, Vinod S Hegade, Rebecca Jones, Simon Rushbrook, Douglas Thorburn, Steve Ryder, Gideon Hirschfield, Jessica K Dyson, David E J Jones. The Serum Proteome and Ursodeoxycholic Acid Response in Primary Biliary Cholangitis. Hepatology (Baltimore, Md.). 2021 12; 74(6):3269-3283. doi: 10.1002/hep.32011. [PMID: 34129689]
  • Gina M Gallucci, Jocelyn Trottier, Christopher Hemme, David N Assis, James L Boyer, Olivier Barbier, Nisanne S Ghonem. Adjunct Fenofibrate Up-regulates Bile Acid Glucuronidation and Improves Treatment Response For Patients With Cholestasis. Hepatology communications. 2021 12; 5(12):2035-2051. doi: 10.1002/hep4.1787. [PMID: 34558841]
  • Ya-Wen Zhang, Ling-Lan Tu, Yi Zhang, Jie-Chao Pan, Gao-Li Zheng, Li-Na Yin. Liver-targeted delivery of asiatic acid nanostructured lipid carrier for the treatment of liver fibrosis. Drug delivery. 2021 Dec; 28(1):2534-2547. doi: 10.1080/10717544.2021.2008054. [PMID: 34854788]
  • Francisco Javier Rodal Canales, Laura Pérez-Campos Mayoral, María Teresa Hernández-Huerta, Luis Manuel Sánchez Navarro, Carlos Alberto Matias-Cervantes, Margarito Martínez Cruz, Eli Cruz Parada, Edgar Zenteno, Edgar Gustavo Ramos-Martínez, Eduardo Pérez-Campos Mayoral, Carlos Romero Díaz, Eduardo Pérez-Campos. Interaction of Spike protein and lipid membrane of SARS-CoV-2 with Ursodeoxycholic acid, an in-silico analysis. Scientific reports. 2021 11; 11(1):22288. doi: 10.1038/s41598-021-01705-5. [PMID: 34782703]
  • Woojae Hong, Jeon Hwang-Bo, Hyelin Jeon, Minsung Ko, Joongyeon Choi, Yong-Joon Jeong, Jae-Hyun Park, Inhye Kim, Tae-Woo Kim, Hyunggun Kim, Se-Chan Kang. A Comparative Study of the Hepatoprotective Effect of Centella asiatica Extract (CA-HE50) on Lipopolysaccharide/d-galactosamine-Induced Acute Liver Injury in C57BL/6 Mice. Nutrients. 2021 Nov; 13(11):. doi: 10.3390/nu13114090. [PMID: 34836346]
  • Aldo J Montano-Loza, Christophe Corpechot. Definition and Management of Patients With Primary Biliary Cholangitis and an Incomplete Response to Therapy. Clinical gastroenterology and hepatology : the official clinical practice journal of the American Gastroenterological Association. 2021 11; 19(11):2241-2251.e1. doi: 10.1016/j.cgh.2020.06.062. [PMID: 32629125]
  • Ergün Sönmezgöz, Sahin Takci, Ali Gül, Murat Uysal. Ursodeoxycholic acid protects neonatal rats from necrotizing enterocolitis: a biochemical, histopathological, and immunohistochemical study. The journal of maternal-fetal & neonatal medicine : the official journal of the European Association of Perinatal Medicine, the Federation of Asia and Oceania Perinatal Societies, the International Society of Perinatal Obstetricians. 2021 Nov; 34(22):3761-3767. doi: 10.1080/14767058.2020.1818210. [PMID: 32954879]
  • Kazufumi Dohmen, Shin-Ya Onohara, Shigeru Harada. Effects of Switching from Fenofibrate to Pemafibrate for Asymptomatic Primary Biliary Cholangitis. The Korean journal of gastroenterology = Taehan Sohwagi Hakhoe chi. 2021 10; 78(4):227-234. doi: 10.4166/kjg.2021.092. [PMID: 34697277]
  • Xuan Qin, Yuanjin Zhang, Jian Lu, Shengbo Huang, Zongjun Liu, Xin Wang. CYP3A deficiency alters bile acid homeostasis and leads to changes in hepatic susceptibility in rats. Toxicology and applied pharmacology. 2021 10; 429(?):115703. doi: 10.1016/j.taap.2021.115703. [PMID: 34461081]
  • Xing-Ling Chen, Shu-Lan Su, Rui Liu, Da-Wei Qian, Li-Ling Chen, Li-Ping Qiu, Jin-Ao Duan. [Chemical constituents and pharmacological action of bile acids from animal:a review]. Zhongguo Zhong yao za zhi = Zhongguo zhongyao zazhi = China journal of Chinese materia medica. 2021 Oct; 46(19):4898-4906. doi: 10.19540/j.cnki.cjcmm.20210630.201. [PMID: 34738383]
  • Yiling ShenTu, Xiaoxiao Mi, Dong Tang, Yanming Jiang, Ling Gao, Xiaojie Ma, Bing Zhou, Wenjun Yang, Junping Shi, Dixiang Lan, Gongying Chen, Ling Gong. Alagille syndrome caused by NOTCH2 mutation presented atypical pathological changes. Clinica chimica acta; international journal of clinical chemistry. 2021 Oct; 521(?):258-263. doi: 10.1016/j.cca.2021.07.026. [PMID: 34332988]
  • Michelle L Manni, Victoria A Heinrich, Gregory J Buchan, James P O'Brien, Crystal Uvalle, Veronika Cechova, Adolf Koudelka, Dharti Ukani, Mohamad Rawas-Qalaji, Tim D Oury, Renee Hart, Madeline Ellgass, Steven J Mullett, Merritt L Fajt, Sally E Wenzel, Fernando Holguin, Bruce A Freeman, Stacy G Wendell. Nitroalkene fatty acids modulate bile acid metabolism and lung function in obese asthma. Scientific reports. 2021 09; 11(1):17788. doi: 10.1038/s41598-021-96471-9. [PMID: 34493738]
  • Hyo Won Jung, Ji Hye Hwang. Anticancer Effects of Ursi Fel Extract and Its Active Compound, Ursodeoxycholic Acid, in FRO Anaplastic Thyroid Cancer Cells. Molecules (Basel, Switzerland). 2021 Sep; 26(17):. doi: 10.3390/molecules26175309. [PMID: 34500742]
  • Fausto Quintavalla, Elisa Gelsi, Luca Battaglia, Raffaella Aldigeri, Roberto Ramoni. Mefepronic acid is associated with a decrease in serum liver enzyme activities in dogs with suspected hepatopathy. Veterinary medicine and science. 2021 09; 7(5):1542-1550. doi: 10.1002/vms3.521. [PMID: 33955681]
  • Valentina Guatibonza-García, Paula Valentina Gaete, Agustín Pérez-Londoño, Danna Kathalina Puerto-Baracaldo, Sebastián Antonio Gutiérrez-Romero, Carlos O Mendivil, Monica Tapias. Poor performance of anti-mitochondrial antibodies for the diagnosis of primary biliary cholangitis in female Colombian patients: A single-center study. World journal of gastroenterology. 2021 Aug; 27(29):4890-4899. doi: 10.3748/wjg.v27.i29.4890. [PMID: 34447233]
  • Wang Chen, Daihua Hu, Zili Feng, Zhaopeng Liu. An effective synthesis of ursodeoxycholic acid from dehydroepiandrosterone. Steroids. 2021 08; 172(?):108870. doi: 10.1016/j.steroids.2021.108870. [PMID: 34038744]
  • Lele Cheng, Tao Chen, Manyun Guo, Peining Liu, Xiangrui Qiao, Yuanyuan Wei, Jianqing She, Bolin Li, Wen Xi, Juan Zhou, Zuyi Yuan, Yue Wu, Junhui Liu. Glycoursodeoxycholic acid ameliorates diet-induced metabolic disorders with inhibiting endoplasmic reticulum stress. Clinical science (London, England : 1979). 2021 07; 135(14):1689-1706. doi: 10.1042/cs20210198. [PMID: 34236076]
  • Nan Shen, Jielu Pan, Hongyu Miao, Haiyan Zhang, Lianjun Xing, Xiao Yu. Fibrates for the treatment of pruritus in primary biliary cholangitis: a systematic review and meta-analysis. Annals of palliative medicine. 2021 Jul; 10(7):7697-7705. doi: 10.21037/apm-21-1304. [PMID: 34353058]
  • Lori W E van der Schoor, Henkjan J Verkade, Anna Bertolini, Sanne de Wit, Elvira Mennillo, Eva Rettenmeier, André A Weber, Rick Havinga, Petra Valášková, Jana Jašprová, Dicky Struik, Vincent W Bloks, Shujuan Chen, Andrea B Schreuder, Libor Vítek, Robert H Tukey, Johan W Jonker. Potential of therapeutic bile acids in the treatment of neonatal Hyperbilirubinemia. Scientific reports. 2021 05; 11(1):11107. doi: 10.1038/s41598-021-90687-5. [PMID: 34045606]
  • Lucas Zangerolamo, Jean F Vettorazzi, Lucas R O Rosa, Everardo M Carneiro, Helena C L Barbosa. The bile acid TUDCA and neurodegenerative disorders: An overview. Life sciences. 2021 May; 272(?):119252. doi: 10.1016/j.lfs.2021.119252. [PMID: 33636170]
  • Lulu Sun, Jie Cai, Frank J Gonzalez. The role of farnesoid X receptor in metabolic diseases, and gastrointestinal and liver cancer. Nature reviews. Gastroenterology & hepatology. 2021 05; 18(5):335-347. doi: 10.1038/s41575-020-00404-2. [PMID: 33568795]
  • Tharni Vasavan, Sahil Deepak, Indu Asanka Jayawardane, Maristella Lucchini, Catherine Martin, Victoria Geenes, Joel Yang, Anita Lövgren-Sandblom, Paul Townsend Seed, Jenny Chambers, Sophia Stone, Lesia Kurlak, Peter Hendy Dixon, Hanns-Ulrich Marschall, Julia Gorelik, Lucy Chappell, Pam Loughna, Jim Thornton, Fiona Broughton Pipkin, Barrie Hayes-Gill, William Paul Fifer, Catherine Williamson. Fetal cardiac dysfunction in intrahepatic cholestasis of pregnancy is associated with elevated serum bile acid concentrations. Journal of hepatology. 2021 05; 74(5):1087-1096. doi: 10.1016/j.jhep.2020.11.038. [PMID: 33276032]
  • Azam Letafat Farashbandi, Mehrdad Shariati, Mokhtar Mokhtari. Comparing the Protective Effects of Curcumin and Ursodeoxycholic Acid after Ethanol-Induced Hepatotoxicity in Rat Liver. Ethiopian journal of health sciences. 2021 May; 31(3):673-682. doi: 10.4314/ejhs.v31i3.25. [PMID: 34483625]
  • Jianliang Xu, P Jaya Kausalya, Noémi Van Hul, Matias J Caldez, Shiyi Xu, Alicia Ghia Min Ong, Wan Lu Woo, Safiah Mohamed Ali, Philipp Kaldis, Walter Hunziker. Protective Functions of ZO-2/Tjp2 Expressed in Hepatocytes and Cholangiocytes Against Liver Injury and Cholestasis. Gastroenterology. 2021 05; 160(6):2103-2118. doi: 10.1053/j.gastro.2021.01.027. [PMID: 33465371]
  • Kan Huang, Chenshu Liu, Meixiu Peng, Qiao Su, Ruiming Liu, Zeling Guo, Sifan Chen, Zilun Li, Guangqi Chang. Glycoursodeoxycholic Acid Ameliorates Atherosclerosis and Alters Gut Microbiota in Apolipoprotein E-Deficient Mice. Journal of the American Heart Association. 2021 04; 10(7):e019820. doi: 10.1161/jaha.120.019820. [PMID: 33787322]
  • Rachida Amzal, Alice Thébaut, Martine Lapalus, Marion Almes, Brigitte Grosse, Elodie Mareux, Mauricette Collado-Hilly, Anne Davit-Spraul, Laure Bidou, Olivier Namy, Emmanuel Jacquemin, Emmanuel Gonzales. Pharmacological Premature Termination Codon Readthrough of ABCB11 in Bile Salt Export Pump Deficiency: An In Vitro Study. Hepatology (Baltimore, Md.). 2021 04; 73(4):1449-1463. doi: 10.1002/hep.31476. [PMID: 32702170]
  • Naruhiro Kimura, Masaaki Takamura, Nobutaka Takeda, Yusuke Watanabe, Yoshihisa Arao, Masahumi Takatsuna, Suguru Takeuchi, Hiroyuki Abe, Toru Setsu, Hiroteru Kamimura, Akira Sakamaki, Kenya Kamimura, Atsunori Tsuchiya, Shuji Terai. Paris II and Rotterdam criteria are the best predictors of outcomes in patients with primary biliary cholangitis in Japan. Hepatology international. 2021 Apr; 15(2):437-443. doi: 10.1007/s12072-021-10163-0. [PMID: 33861397]
  • Akash Roy, Madhumita Premkumar, Saurabh Mishra, Rohit Mehtani, Vanita Suri, Neelam Aggarwal, Surender Singh, Radha Krishna Dhiman. Role of ursodeoxycholic acid on maternal serum bile acids and perinatal outcomes in intrahepatic cholestasis of pregnancy. European journal of gastroenterology & hepatology. 2021 04; 33(4):571-576. doi: 10.1097/meg.0000000000001954. [PMID: 33136720]
  • Hélène Céruti, Gilles Kayem, Lucie Guilbaud, Chloé Dussaux, Anne Gervais, Aurélie Beaufrère, Benoit Coffin, Laurent Mandelbrot, Emeline Maisonneuve. Intrahepatic cholestasis of pregnancy associated with azathioprine: A case series. Journal of gynecology obstetrics and human reproduction. 2021 Apr; 50(4):102083. doi: 10.1016/j.jogoh.2021.102083. [PMID: 33601075]
  • Maria Nadinskaia, Marina Maevskaya, Vladimir Ivashkin, Khava Kodzoeva, Irina Pirogova, Evgeny Chesnokov, Alexander Nersesov, Jamilya Kaibullayeva, Akzhan Konysbekova, Aigul Raissova, Feruza Khamrabaeva, Elena Zueva. Ursodeoxycholic acid as a means of preventing atherosclerosis, steatosis and liver fibrosis in patients with nonalcoholic fatty liver disease. World journal of gastroenterology. 2021 Mar; 27(10):959-975. doi: 10.3748/wjg.v27.i10.959. [PMID: 33776366]
  • Ye Sirchak, M Derbak, M Stan, O Petrichko. INFLUENCE OF URSODEOXYCHOLIC ACID ON THE CHOLECYSTOKININ LEVELS IN PATIENTS WITH GASTRO-ESOPHAGEAL REFLUX DISEASE AND TYPE 2 DIABETES MELLITUS. Georgian medical news. 2021 Mar; ?(312):67-71. doi: . [PMID: 33964829]
  • Debashis Haldar, Ashnila Janmohamed, Tim Plant, Matthew Davidson, Hannah Norman, Emily Russell, Olivia Serevina, Kenneth Chung, Kashif Qamar, Bridget Gunson, Bettina Hansen, Alex Richter, Palak J Trivedi, Gideon M Hirschfield. Antibodies to gp210 and understanding risk in patients with primary biliary cholangitis. Liver international : official journal of the International Association for the Study of the Liver. 2021 03; 41(3):535-544. doi: 10.1111/liv.14688. [PMID: 33022821]
  • Xi Dong, Yun Luo, Shan Lu, Han Ma, Wenchao Zhang, Yue Zhu, Guibo Sun, Xiaobo Sun. Ursodesoxycholic acid alleviates liver fibrosis via proregeneration by activation of the ID1-WNT2/HGF signaling pathway. Clinical and translational medicine. 2021 02; 11(2):e296. doi: 10.1002/ctm2.296. [PMID: 33635004]
  • Richard H Lee, Mara Greenberg, Torri D Metz, Christian M Pettker. Society for Maternal-Fetal Medicine Consult Series #53: Intrahepatic cholestasis of pregnancy: Replaces Consult #13, April 2011. American journal of obstetrics and gynecology. 2021 02; 224(2):B2-B9. doi: 10.1016/j.ajog.2020.11.002. [PMID: 33197417]
  • Christine Wu, Maria-Elisabeth Smet, Susan Heath, Jennifer Curnow, Ming-Wei Lin, Jacob George, Thushari I Alahakoon. Pregnancy complicated by refractory severe hypercholanaemia from sodium taurocholate co-transporting polypeptide deficiency. The journal of obstetrics and gynaecology research. 2021 Feb; 47(2):822-826. doi: 10.1111/jog.14568. [PMID: 33174277]
  • Gilda Porta, Elisa de Carvalho, Jorge L Santos, Jorge Gama, Jorge A Bezerra. Autoimmune Hepatitis: Predictors of Native Liver Survival in Children and Adolescents. The Journal of pediatrics. 2021 02; 229(?):95-101.e3. doi: 10.1016/j.jpeds.2020.10.009. [PMID: 33500120]
  • Yuan Gao, Li Li, Bei Li, Yutao Zhan. Response Rate and Impact on Lipid Profiles of Obeticholic Acid Treatment for Patients with Primary Biliary Cholangitis: A Meta-Analysis. Canadian journal of gastroenterology & hepatology. 2021; 2021(?):8829510. doi: 10.1155/2021/8829510. [PMID: 33511089]
  • Jenessa A Winston, Alissa Rivera, Jingwei Cai, Andrew D Patterson, Casey M Theriot. Secondary bile acid ursodeoxycholic acid alters weight, the gut microbiota, and the bile acid pool in conventional mice. PloS one. 2021; 16(2):e0246161. doi: 10.1371/journal.pone.0246161. [PMID: 33600468]
  • Jan Freark de Boer, Hilde D de Vries, Anna Palmiotti, Rumei Li, Marwah Doestzada, Joanne A Hoogerland, Jingyuan Fu, Anouk M La Rose, Marit Westerterp, Niels L Mulder, Milaine V Hovingh, Martijn Koehorst, Niels J Kloosterhuis, Justina C Wolters, Vincent W Bloks, Joel T Haas, David Dombrowicz, Bart Staels, Bart van de Sluis, Folkert Kuipers. Cholangiopathy and Biliary Fibrosis in Cyp2c70-Deficient Mice Are Fully Reversed by Ursodeoxycholic Acid. Cellular and molecular gastroenterology and hepatology. 2021; 11(4):1045-1069. doi: 10.1016/j.jcmgh.2020.12.004. [PMID: 33309945]
  • Seonghae Yoon, Heechan Lee, Sang-Chun Ji, Seo Hyun Yoon, Joo-Youn Cho, Jae-Yong Chung. Pharmacokinetics and Pharmacodynamics of Ursodeoxycholic Acid in an Overweight Population With Abnormal Liver Function. Clinical pharmacology in drug development. 2021 01; 10(1):68-77. doi: 10.1002/cpdd.790. [PMID: 32191400]
  • Masaaki Hirayama, Hiroshi Nishiwaki, Tomonari Hamaguchi, Mikako Ito, Jun Ueyama, Tetsuya Maeda, Kenichi Kashihara, Yoshio Tsuboi, Kinji Ohno. Intestinal Collinsella may mitigate infection and exacerbation of COVID-19 by producing ursodeoxycholate. PloS one. 2021; 16(11):e0260451. doi: 10.1371/journal.pone.0260451. [PMID: 34813629]
  • Nisanne S Ghonem, Adam M Auclair, Christopher L Hemme, Gina M Gallucci, Randolph de la Rosa Rodriguez, James L Boyer, David N Assis. Fenofibrate Improves Liver Function and Reduces the Toxicity of the Bile Acid Pool in Patients With Primary Biliary Cholangitis and Primary Sclerosing Cholangitis Who Are Partial Responders to Ursodiol. Clinical pharmacology and therapeutics. 2020 12; 108(6):1213-1223. doi: 10.1002/cpt.1930. [PMID: 32480421]
  • Brittany C Kunz, Sharon A Center, John F Randolph, Janelle D Walker, April E Choi, Karl E Anderson. Congenital erythropoietic protoporphyria and protoporphyric hepatopathy in a dog. Journal of the American Veterinary Medical Association. 2020 Dec; 257(11):1148-1156. doi: 10.2460/javma.2020.257.11.1148. [PMID: 33226294]
  • Xing Chen, Xiao Ma, Ruilin Wang, Lifu Wang, Jianyu Li, Honghong Liu, Tingting He, Shizhang Wei, Haotian Li, Min Wang, Yanling Zhao. Treatment of primary biliary cirrhosis with ursodeoxycholic acid combined with traditional Chinese medicine: A protocol for systematic review and meta analysis. Medicine. 2020 Nov; 99(46):e23107. doi: 10.1097/md.0000000000023107. [PMID: 33181677]
  • Adrian M Di Bisceglie, Gerald F Watts, Philip Lavin, Meng Yu, Ru Bai, Liping Liu. Pharmacokinetics and pharmacodynamics of HTD1801 (berberine ursodeoxycholate, BUDCA) in patients with hyperlipidemia. Lipids in health and disease. 2020 Nov; 19(1):239. doi: 10.1186/s12944-020-01406-4. [PMID: 33183320]
  • Kristel K Leung, Maya Deeb, Gideon M Hirschfield. Review article: pathophysiology and management of primary biliary cholangitis. Alimentary pharmacology & therapeutics. 2020 10; 52(7):1150-1164. doi: 10.1111/apt.16023. [PMID: 32813299]
  • Saleem Abdulrab, Sadeq Al-Maweri, Esam Halboub. Ursodeoxycholic acid as a candidate therapeutic to alleviate and/or prevent COVID-19-associated cytokine storm. Medical hypotheses. 2020 10; 143(?):109897. doi: 10.1016/j.mehy.2020.109897. [PMID: 32505909]
  • Iván L Csanaky, Andrew J Lickteig, Youcai Zhang, Curtis D Klaassen. Effects of patent ductus venosus on bile acid homeostasis in aryl hydrocarbon receptor (AhR)-null mice. Toxicology and applied pharmacology. 2020 09; 403(?):115136. doi: 10.1016/j.taap.2020.115136. [PMID: 32679164]
  • Manuela R Martinefski, Myrian R Rodriguez, Fabián Buontempo, Silvia E Lucangioli, Liliana G Bianciotti, Valeria P Tripodi. Coenzyme Q 10 supplementation: A potential therapeutic option for the treatment of intrahepatic cholestasis of pregnancy. European journal of pharmacology. 2020 Sep; 882(?):173270. doi: 10.1016/j.ejphar.2020.173270. [PMID: 32534074]
  • Xuan Li, Min Liao, Qiong Pan, Qiaoling Xie, Hong Yang, Ying Peng, Qiao Li, Jiaquan Qu, Jin Chai. Combination therapy of obeticholic acid and ursodeoxycholic acid in patients with primary biliary cholangitis who respond incompletely to ursodeoxycholic acid: a systematic review. European journal of gastroenterology & hepatology. 2020 09; 32(9):1116-1122. doi: 10.1097/meg.0000000000001785. [PMID: 32649329]
  • Jiani Huang, Fang Wen, Wenjie Huang, Yingfeng Bai, Xiaona Lu, Peng Shu. Identification of hub genes and discovery of promising compounds in gastric cancer based on bioinformatics analysis. Biomarkers in medicine. 2020 08; 14(12):1069-1084. doi: 10.2217/bmm-2019-0608. [PMID: 32969243]
  • Mario Simental-Mendía, Adriana Sánchez-García, Luis E Simental-Mendía. Effect of ursodeoxycholic acid on liver markers: A systematic review and meta-analysis of randomized placebo-controlled clinical trials. British journal of clinical pharmacology. 2020 08; 86(8):1476-1488. doi: 10.1111/bcp.14311. [PMID: 32285958]
  • Hui Yang, Hang Yang, Lixia Wang, Honggang Shi, Bojia Liu, Xue Lin, Qingyong Chang, Jiande D Z Chen, Zhijun Duan. Transcutaneous Neuromodulation improved inflammation and sympathovagal ratio in patients with primary biliary ssscholangitis and inadequate response to Ursodeoxycholic acid: a pilot study. BMC complementary medicine and therapies. 2020 Aug; 20(1):242. doi: 10.1186/s12906-020-03036-w. [PMID: 32738911]
  • Kate F Walker, Lucy C Chappell, William M Hague, Philippa Middleton, Jim G Thornton. Pharmacological interventions for treating intrahepatic cholestasis of pregnancy. The Cochrane database of systematic reviews. 2020 07; 7(?):CD000493. doi: 10.1002/14651858.cd000493.pub3. [PMID: 32716060]
  • Ning Fan, Ke Meng, Yuqing Zhang, Yong Hu, Donghua Li, Qiaoying Gao, Jianhua Wang, Yanning Li, Shangwei Wu, Yunfeng Cui. The effect of ursodeoxycholic acid on the relative expression of the lipid metabolism genes in mouse cholesterol gallstone models. Lipids in health and disease. 2020 Jul; 19(1):158. doi: 10.1186/s12944-020-01334-3. [PMID: 32615989]
  • Luiza Borges Manna, Georgia Papacleovoulou, Flavia Flaviani, Vanessa Pataia, Asaad Qadri, Shadi Abu-Hayyeh, Saraid McIlvride, Eugene Jansen, Peter Dixon, Jennifer Chambers, Marta Vazquez-Lopez, Annika Wahlström, Negusse Kitaba, Hanns-Ulrich Marschall, Keith M Godfrey, Karen Lillycrop, Catherine Williamson. Ursodeoxycholic acid improves feto-placental and offspring metabolic outcomes in hypercholanemic pregnancy. Scientific reports. 2020 06; 10(1):10361. doi: 10.1038/s41598-020-67301-1. [PMID: 32587408]
  • Qing Xiao, Shujun Zhang, Huina Ren, Ruoyang Du, Jiajun Li, Jinqiu Zhao, Yue Gao, Yali Zhu, Wenxiang Huang. Ginsenoside Rg1 alleviates ANIT-induced intrahepatic cholestasis in rats via activating farnesoid X receptor and regulating transporters and metabolic enzymes. Chemico-biological interactions. 2020 Jun; 324(?):109062. doi: 10.1016/j.cbi.2020.109062. [PMID: 32198087]
  • Başak Gümüş Güler, Sibel Özler. Increased syndecan-1 and glypican-3 predict poor perinatal outcome and treatment resistance in intrahepatic cholestasis. Hepatobiliary & pancreatic diseases international : HBPD INT. 2020 Jun; 19(3):271-276. doi: 10.1016/j.hbpd.2019.12.001. [PMID: 31919038]
  • Abhishek G Sathe, Paul Tuite, Chi Chen, Yiwei Ma, Wei Chen, James Cloyd, Walter C Low, Clifford J Steer, Byeong-Yeul Lee, Xiao-Hong Zhu, Lisa D Coles. Pharmacokinetics, Safety, and Tolerability of Orally Administered Ursodeoxycholic Acid in Patients With Parkinson's Disease-A Pilot Study. Journal of clinical pharmacology. 2020 06; 60(6):744-750. doi: 10.1002/jcph.1575. [PMID: 32052462]
  • Patryk Lipiński, Dorota Jurkiewicz, Elżbieta Ciara, Rafał Płoski, Sabina Więcek, Anna Bogdańska, Teresa Stradomska, Piotr Socha, Dariusz Rokicki, Anna Tylki-Szymańska, Irena Jankowska. Neonatal cholestasis due to citrin deficiency: diagnostic pitfalls. Acta biochimica Polonica. 2020 May; 67(2):225-228. doi: 10.18388/abp.2020_5202. [PMID: 32436673]
  • Yunwen Yang, Suwen Liu, Huiping Gao, Peipei Wang, Yue Zhang, Aihua Zhang, Zhanjun Jia, Songming Huang. Ursodeoxycholic acid protects against cisplatin-induced acute kidney injury and mitochondrial dysfunction through acting on ALDH1L2. Free radical biology & medicine. 2020 05; 152(?):821-837. doi: 10.1016/j.freeradbiomed.2020.01.182. [PMID: 32004633]
  • Edilson Ribeiro de Oliveira Junior, Eleonora Truzzi, Luca Ferraro, Marco Fogagnolo, Barbara Pavan, Sarah Beggiato, Cecilia Rustichelli, Eleonora Maretti, Eliana Martins Lima, Eliana Leo, Alessandro Dalpiaz. Nasal administration of nanoencapsulated geraniol/ursodeoxycholic acid conjugate: Towards a new approach for the management of Parkinson's disease. Journal of controlled release : official journal of the Controlled Release Society. 2020 05; 321(?):540-552. doi: 10.1016/j.jconrel.2020.02.033. [PMID: 32092370]
  • Jie Wang, Xiang-Zhong Gu, Li-Ming He, Chen-Chen Li, Wen-Wei Qiu. Synthesis of ursodeoxycholic acid from plant-source (20S)-21-hydroxy-20-methylpregn-4-en-3-one. Steroids. 2020 05; 157(?):108600. doi: 10.1016/j.steroids.2020.108600. [PMID: 32068080]